...
首页> 外文期刊>Stem cells international >Potential Effect of Exosomes Derived from Cancer Stem Cells and MSCs on Progression of DEN-Induced HCC in Rats
【24h】

Potential Effect of Exosomes Derived from Cancer Stem Cells and MSCs on Progression of DEN-Induced HCC in Rats

机译:癌症干细胞和MSCs外来体对大鼠DEN诱导的HCC进程的潜在影响

获取原文

摘要

Cross talk, mediated by exosomes, between normal stem cells and cancer stem cells (CSCs) in the tumor microenvironment has been given less attention so far. In addition, no publications are available in the literature that address the in vivo impact of exosomes derived from CSCs and mesenchymal stem cells (MSCs) on progression of long-term hepatocellular carcinoma (HCC). Herein, we hypothesized that transfer of exosomes among the cells in the HCC microenvironment could either induce or inhibit tumor growth and metastasis depending on their source. To check this hypothesis, we investigated the effect of exosomes coming from two different stem cell populations, hepatic CSCs and bone marrow (BM) MSCs, on progression of long-term DEN-induced HCC in rats and the involved underlying mechanisms. CSCs-exosomes induced a significant increase in liver relative weight and serum levels of cancer markers (AFP and GGT) and liver enzymes (ALT, AST, and ALP), intensive immunostaining for the HCC marker GST-P, and an increased number and area of tumor nodules as compared to HCC rats injected by PBS. CSCs-exosomes also decreased apoptosis (marked by downregulation of Bax and p53 and upregulation of Bcl2, and increased immunostaining of PCNA), increased angiogenetic activity (revealed by upregulation of VEGF), enhanced metastasis and invasiveness (indicated by upregulation of P13K and ERK proteins and their downstream target MMP9 and downregulation of TIMP1), and induced epithelial mesenchymal transition (marked by increased serum and hepatic level of TGFβ1 mRNA and protein). Notably, CSCs-exosomes also elevated HCC exosomal microRNA (miR) 21, exosomal long noncoding (lnc) RNA Tuc339, lncHEIH, and the HCC lncHOTAIR and decreased liver miR122 and HCC miRs (miR148a, miR16, and miR125b). All these cellular, functional, and molecular changes were reversed following injection of BM-MSCs-exosomes. However, both CSCs- and MSCs-exosomes failed to change the elevated oxidative stress or the inhibited antioxidant activities induced by HCC. Collectively, our results revealed a tumor stimulatory effect (induction of tumor growth, progression, and metastasis) for exosomes derived from CSCs and an inhibitory effect for exosomes derived from MSCs. These results provide valuable insight on the effect of CSCs- and MSCs-exosomes on HCC growth and progression in vivo, which may be helpful to understand the mechanism of HCC development.
机译:迄今为止,肿瘤微环境中正常干细胞与癌干细胞(CSC)之间由外来体介导的串扰受到的关注较少。另外,在文献中没有出版物涉及源自CSC和间充质干细胞(MSC)的外来体对长期肝细胞癌(HCC)进展的体内影响。本文中,我们假设外泌体在HCC微环境中的细胞之间转移可能会诱导或抑制肿瘤的生长和转移,具体取决于其来源。为了检验这一假设,我们研究了来自两种不同干细胞群体(肝CSC和骨髓(BM)MSC)的外泌体对大鼠长期DEN诱导的HCC进程的影响以及所涉及的潜在机制。 CSCs外泌体显着增加了肝脏相对重量和癌症标志物(AFP和GGT)和肝酶(ALT,AST和ALP)的血清水平,对HCC标志物GST-P的强化免疫染色以及数量和面积的增加与通过PBS注射的HCC大鼠相比,肿瘤结节的数量增加。 CSCs外泌体还减少凋亡(以Bax和p53的下调和Bcl2的上调为标志,并增加PCNA的免疫染色),增加的血管生成活性(通过VEGF的上调来证明),增强的转移和侵袭性(以P13K和ERK蛋白的上调表示)以及它们的下游靶标MMP9和TIMP1的下调),并诱导上皮间质转化(以血清和肝中TGFβ1mRNA和蛋白质的水平升高为标志)。值得注意的是,CSCs外来体还升高了HCC外泌体microRNA(miR)21,外泌体长非编码(lnc)RNA Tuc339,lncHEIH和HCC lncHOTAIR,并降低了肝miR122和HCC miRs(miR148a,miR16和miR125b)。注射BM-MSCs-外泌体后,所有这些细胞,功能和分子变化均被逆转。然而,CSCs和MSCs外泌体均不能改变HCC诱导的氧化应激升高或抗氧化活性降低。总体而言,我们的研究结果显示,源自CSC的外泌体具有肿瘤刺激作用(诱导肿瘤生长,进展和转移),而源自MSC的外泌体则具有抑制作用。这些结果为CSCs和MSCs外来体对体内HCC生长和进展的作用提供了有价值的见解,这可能有助于了解HCC发生的机制。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号